Elsevier

Biomaterials

Volume 196, March 2019, Pages 80-89
Biomaterials

Mesenchymal stem cell-macrophage crosstalk and bone healing

https://doi.org/10.1016/j.biomaterials.2017.12.025Get rights and content

Abstract

Recent research has brought about a clear understanding that successful fracture healing is based on carefully coordinated cross-talk between inflammatory and bone forming cells. In particular, the key role that macrophages play in the recruitment and regulation of the differentiation of mesenchymal stem cells (MSCs) during bone regeneration has been brought to focus. Indeed, animal studies have comprehensively demonstrated that fractures do not heal without the direct involvement of macrophages. Yet the exact mechanisms by which macrophages contribute to bone regeneration remain to be elucidated. Macrophage–derived paracrine signaling molecules such as Oncostatin M, Prostaglandin E2 (PGE2), and Bone Morphogenetic Protein-2 (BMP2) have been shown to play critical roles; however the relative importance of inflammatory (M1) and tissue regenerative (M2) macrophages in guiding MSC differentiation along the osteogenic pathway remains poorly understood. In this review, we summarize the current understanding of the interaction of macrophages and MSCs during bone regeneration, with the emphasis on the role of macrophages in regulating bone formation. The potential implications of aging to this cellular cross-talk are reviewed. Emerging treatment options to improve facture healing by utilizing or targeting MSC-macrophage crosstalk are also discussed.

Introduction

Bone fractures are one of the most common injuries seen in emergency departments, with nearly 4 million fractures seen in the United States in 2013 [1]. Despite the best treatment efforts, up to 10% of bone fracture cases still report undesirable outcomes; in the USA alone, 100,000 fractures per year result in painful non-union [2]. Treatment of these non-united fractures and bone defects constitutes a major health problem with significant clinical, social, and economic implications with an average cost of over 10,000 USD per non-union [3].

There are two major pathways for bone regeneration: intramembranous or endochondral ossification processes [[4], [5], [6], [7]]. Intramembranous ossification involves mesenchymal stem cells (MSCs) directly differentiating into osteoblasts which in turn deposit mineralized extracellular matrix. This type of healing is typically seen in rigidly fixed fractures with minimal fracture gap, and with fractures within the bone metaphysis. Fractures located in the diaphysis, with less mechanical stability, and a larger fracture gap heal via the classic stages of endochondral ossification: inflammation, soft then hard callus formation, and finally remodeling of the fracture site. In this mode of bone regeneration, the fracture hematoma is initially infiltrated by immune cells, mainly neutrophils and macrophages. Macrophages not only phagocytose necrotic cells and tissue debris at the fracture site but also initiate the recruitment of MSCs and vascular progenitor cells from the periosteum, bone marrow, and circulation [8,9]. As the inflammation subsides, MSCs and other progenitor cells proliferate, forming granulation tissue that ultimately forms cartilage callus to stabilize the fracture site [7,10]. In addition to providing mechanical stability cartilage functions as a scaffold for osteoblast-mediated bone deposition that allows for mineralization of the callus and closure of the fracture gap [11,12]. Osteoclasts then resorb immature woven bone and cartilage matrix, and with the subsequent deposition of mature lamellar bone, bone is restored to its pre-fracture structure and integrity [7,10].

The differentiation of MSCs and the subsequent formation of cartilage and bone at the fracture site is guided by several microenvironmental signals. These include growth factors released from the bone matrix as well as changes in the oxygen tension and mechanical microenvironment [7,10]. In particular, recent research has determined that both routes of fracture healing are based on carefully coordinated cross-talk between macrophages and bone forming cells.

MSCs, the precursor cells for bone and cartilage, were initially identified from human bone marrow with the ability to develop into fibroblastic colony-forming cells in vitro, and to regenerate heterotopic bone tissue in vivo [13,14]. The exact definition of the MSC remains controversial, but the term is generally used to describe a population of stem cells that resides in the peri-vascular niche of most tissues, and with the ability to differentiate into mesodermal tissues, such as bone and cartilage [15,16]. The International Society for Cellular Therapy has defined MSCs as cells that 1) adhere to plastic in vitro cell cultures; 2) have a certain surface marker profile (CD105+, CD73+, CD90+, and CD45−, CD34−, CD14−, CD11b−, CD79−, CD19−, and HLA-DR-); and 3) have the trilineage ability to differentiate into osteoblasts, adipocytes, and chondroblasts [17]. In addition to their ability to regenerate mesenchymal tissues, MSCs have wide immunomodulatory properties making them attractive targets for tissue engineering applications.

Macrophages are cells of innate immunity that are found in nearly all tissues, where they play a key role in maintaining normal tissue homeostasis [18]. During infection and inflammation, their numbers increase greatly via homing of regional and circulating monocyte precursors to the affected area [19]. During acute inflammation, macrophages contribute to the restoration of tissue homeostasis by phagocytosing invading micro-organisms, amplifying the inflammatory reaction, and recruiting additional immune cells [20]. As the tissue insult is cleared, macrophages contribute to tissue regeneration by secreting anti-inflammatory factors, recruiting progenitor cells, and producing growth factors that regulate the differentiation of these cells including angiogenesis [18,20]. This functional plasticity of macrophages has been conceptualized as macrophage polarization; inflammatory macrophages are called classically activated or M1 macrophages, while macrophages active in tissue regeneration are known as alternatively activated or M2 macrophages [21]. Importantly macrophages can switch from one mode of function to another, making them highly attractive targets for therapeutic interventions. In humans, there is a much wider spectrum of macrophage phenotypes, corresponding to relative differences in pro- and anti-inflammatory activities.

Macrophages are among the first cells to arrive to the fracture site, and have long been thought to contribute to the initial inflammation and debridement of the injury location (Fig. 1). Their key role also in the regulation of bone regeneration during both normal bone homeostasis and fracture healing has increasingly been appreciated. In addition to macrophages, closely related myeloid-lineage cells such as osteoclasts play complex roles in bone growth and regeneration [[22], [23], [24]], and delineating the exact roles that each of these myeloid lineage cell types play in bone regeneration remains challenging with currently available methods. Nevertheless the research has begun to identify some of the molecular mechanisms underlying the cross-talk between macrophages and bone forming cells and has led to new potential strategies to enhance bone regeneration by targeting the interaction between macrophages and MSCs.

Section snippets

Macrophages in bone homeostasis

Bone tissue contains a resident macrophage subpopulation termed osteomacs, which is distributed among bone lining cells within both endosteum and periosteum [25]. In particular osteomacs are closely associated with areas of bone formation, forming a canopy-like structure over active cuboidal osteoblasts. When macrophages were depleted in a macrophage-fas-induced apoptosis (MAFIA) model [26], this active bone forming surface of osteoblasts was also lost, suggesting an active role for the

Mechanisms of macrophage/MSC interaction

The exact mechanisms by which macrophages contribute to bone regeneration remain unclear. It is likely that the initial inflammatory reaction and pro-inflammatory macrophage activation contributes to the recruitment of MSCs and osteoprogenitor and vascular progenitor cells to the fracture site. These signals that control progenitor cell homing include chemokines CCL2, CXCL8 and SDF-1, all of which are secreted by activated macrophages [[38], [39], [40], [41], [42]]. The general paradigm of

Impact of aging on the MSC/macrophage cross-talk

There is increasing evidence that aging induces profound changes in the physiology of both macrophages and MSCs. Choudhery et al. [70] harvested young and aged murine MSCs and compared their regenerative potential. Through a tube-forming assay with matrigel, the authors showed a decrease in wound healing with aged MSCs, and significant downregulation of VEGF, SDF-1 and protein kinase B. Bernet at al. [71] showed that aged resident muscle MSCs lose their self-renewal abilities via alterations in

Therapeutic potential of utilizing macrophage-MSC crosstalk for bone regeneration

Current research on cell-based bone tissue engineering has largely focused on utilizing the bone regenerative potential of MSCs. [83,84] MSCs are highly attractive targets for cell-based therapies to induce bone formation at the site of the bone defect or non-union, given their multi-potency for bone, cartilage, and blood vessels, and the ease of harvesting MSCs from adult tissues and expanding them in vitro. In contrast, relatively little research has thus far focused on utilizing macrophages

Macrophage polarization, duration of inflammation, and bone formation

Studies using animal models have established that macrophage/MSC cross talk is crucial for bone regeneration and that macrophages likely have the greatest impact on fracture healing during the early inflammatory phase. The mechanisms of interaction of monocyte/macrophages and bone forming cells have been studied in vitro. Although these studies have arrived at the same general conclusion that macrophages promote MSC and pre-osteoblast mediated bone formation, details of the interaction remain

Acknowledgements

This work was supported by NIH grants 2R01AR055650, 1R01AR063717 and the Ellenburg Chair in Surgery at Stanford University. J.P. was supported by a grant from the Jane and Aatos Erkko Foundation.

References (104)

  • L.J. Raggatt et al.

    Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification

    Am. J. Pathol.

    (2014 Dec)
  • I.G. Winkler et al.

    Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs

    Blood

    (2010 Dec 2)
  • F. Belema-Bedada et al.

    Efficient homing of multipotent adult mesenchymal stem cells depends on FROUNT-mediated clustering of CCR2

    Cell Stem Cell

    (2008 Jun 5)
  • A. Mantovani et al.

    The chemokine system in diverse forms of macrophage activation and polarization

    Trends Immunol.

    (2004 Dec)
  • C.M. Champagne et al.

    Macrophage cell lines produce osteoinductive signals that include bone morphogenetic protein-2

    Bone

    (2002 Jan)
  • O.M. Omar et al.

    The stimulation of an osteogenic response by classical monocyte activation

    Biomaterials

    (2011 Nov)
  • N. Fahy et al.

    Human osteoarthritic synovium impacts chondrogenic differentiation of mesenchymal stem cells via macrophage polarisation state

    Osteoarthritis Cartilage

    (2014 Aug)
  • A.J. Nauta et al.

    Immunomodulatory properties of mesenchymal stromal cells

    Blood

    (2007 Nov 15)
  • G. Ren et al.

    Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide

    Cell Stem Cell

    (2008 Feb 7)
  • J. Kim et al.

    Mesenchymal stem cell-educated macrophages: a novel type of alternatively activated macrophages

    Exp. Hematol.

    (2009 Dec)
  • M. Francois et al.

    Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation

    Mol. Ther.

    (2012 Jan)
  • E. Seebach et al.

    Mesenchymal stroma cells trigger early attraction of M1 macrophages and endothelial cells into fibrin hydrogels, stimulating long bone healing without long-term engraftment

    Acta Biomater.

    (2014 Nov)
  • M.E. Bernardo et al.

    Mesenchymal stromal cells: sensors and switchers of inflammation

    Cell Stem Cell

    (2013 Oct 3)
  • S. Lee et al.

    Activated mesenchymal stem cells increase wound tensile strength in aged mouse model via macrophages

    J. Surg. Res.

    (2013 May 1)
  • I. Fernandez-Bances et al.

    Repair of long-bone pseudoarthrosis with autologous bone marrow mononuclear cells combined with allogenic bone graft

    Cytotherapy

    (2013 May)
  • S.B. Goodman et al.

    Treatment of secondary osteonecrosis of the knee with local debridement and osteoprogenitor cell grafting

    J. Arthroplasty

    (2015 Nov)
  • Z. Chen et al.

    Osteogenic differentiation of bone marrow MSCs by beta-tricalcium phosphate stimulating macrophages via BMP2 signalling pathway

    Biomaterials

    (2014 Feb)
  • Z. Chen et al.

    Osteoimmunomodulatory properties of magnesium scaffolds coated with beta-tricalcium phosphate

    Biomaterials

    (2014 Oct)
  • K.L. Spiller et al.

    Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds

    Biomaterials

    (2015 Jan)
  • D. Hachim et al.

    Shifts in macrophage phenotype at the biomaterial interface via IL-4 eluting coatings are associated with improved implant integration

    Biomaterials

    (2017 Jan)
  • T. Lin et al.

    Establishment of NF-kappaB sensing and interleukin-4 secreting mesenchymal stromal cells as an “on-demand” drug delivery system to modulate inflammation

    Cytotherapy

    (2017 Sep)
  • C. Control

    National Hospital Ambulatory Medical Care Survey: 2013 Emergency Department Summary Tables

    (2013)
  • M. Miranda et al.

    Treatment strategy for nonunions and malunions

  • H.K. Uhthoff et al.

    Healing patterns of metaphyseal fractures

    Clin. Orthop. Relat. Res.

    (1981 Oct)
  • P. Aspenberg et al.

    Distal radial fractures heal by direct woven bone formation

    Acta Orthop.

    (2013 Jun)
  • O. Bastian et al.

    Systemic inflammation and fracture healing

    J. Leukoc. Biol.

    (2011 May)
  • L. Claes et al.

    Fracture healing under healthy and inflammatory conditions

    Nat. Rev. Rheumatol.

    (2012 Jan 31)
  • A.J. Friedenstein et al.

    Osteogenesis in transplants of bone marrow cells

    J. Embryol. Exp. Morphol.

    (1966 Dec)
  • A.J. Friedenstein et al.

    Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo

    Transplantation

    (1974 Apr)
  • A. Keating

    Mesenchymal stromal cells

    Curr. Opin. Hematol.

    (2006 Nov)
  • A. Uccelli et al.

    Mesenchymal stem cells in health and disease

    Nat. Rev. Immunol.

    (2008)
  • P.J. Murray et al.

    Protective and pathogenic functions of macrophage subsets

    Nat. Rev. Immunol.

    (2011 Oct 14)
  • C. Shi et al.

    Monocyte recruitment during infection and inflammation

    Nat. Rev. Immunol.

    (2011 Oct 10)
  • D.M. Mosser et al.

    Exploring the full spectrum of macrophage activation

    Nat. Rev. Immunol.

    (2008 Dec)
  • F.O. Martinez et al.

    Macrophage activation and polarization

    Front. Biosci.

    (2008 Jan 1)
  • P.R. Odgren et al.

    The cast of clasts: catabolism and vascular invasion during bone growth, repair, and disease by osteoclasts, chondroclasts, and septoclasts

    Connect. Tissue Res.

    (2016 May)
  • M.K. Chang et al.

    Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo

    J. Immunol.

    (2008 Jul 15)
  • S.H. Burnett et al.

    Conditional macrophage ablation in transgenic mice expressing a Fas-based suicide gene

    J. Leukoc. Biol.

    (2004 Apr)
  • L. Vi et al.

    Macrophages promote osteoblastic differentiation in-vivo: implications in fracture repair and bone homeostasis

    J. Bone Miner. Res.

    (2015 Jun)
  • K.A. Alexander et al.

    Osteal macrophages promote in vivo intramembranous bone healing in a mouse tibial injury model

    J. Bone Miner. Res.

    (2011 Jul)
  • Cited by (556)

    View all citing articles on Scopus
    View full text